Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
FEBS J ; 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38390750

ABSTRACT

The identification of the coproporphyrin-dependent heme biosynthetic pathway, which is used almost exclusively by monoderm bacteria in 2015 by Dailey et al. triggered studies aimed at investigating the enzymes involved in this pathway that were originally assigned to the protoporphyrin-dependent heme biosynthetic pathway. Here, we revisit the active site of coproporphyrin ferrochelatase by a biophysical and biochemical investigation using the physiological substrate coproporphyrin III, which in contrast to the previously used substrate protoporphyrin IX has four propionate substituents and no vinyl groups. In particular, we have compared the reactivity of wild-type coproporphyrin ferrochelatase from the firmicute Listeria monocytogenes with those of variants, namely, His182Ala (H182A) and Glu263Gln (E263Q), involving two key active site residues. Interestingly, both variants are active only toward the physiological substrate coproporphyrin III but inactive toward protoporphyrin IX. In addition, E263 exchange impairs the final oxidation step from ferrous coproheme to ferric coproheme. The characteristics of the active site in the context of the residues involved and the substrate binding properties are discussed here using structural and functional means, providing a further contribution to the deciphering of this enigmatic reaction mechanism.

2.
Protein Sci ; 32(11): e4788, 2023 11.
Article in English | MEDLINE | ID: mdl-37743577

ABSTRACT

Understanding the reaction mechanism of enzymes at the molecular level is generally a difficult task, since many parameters affect the turnover. Often, due to high reactivity and formation of transient species or intermediates, detailed information on enzymatic catalysis is obtained by means of model substrates. Whenever possible, it is essential to confirm a reaction mechanism based on substrate analogues or model systems by using the physiological substrates. Here we disclose the ferrous iron incorporation mechanism, in solution, and in crystallo, by the coproporphyrin III-coproporphyrin ferrochelatase complex from the firmicute, pathogen, and antibiotic resistant, Listeria monocytogenes. Coproporphyrin ferrochelatase plays an important physiological role as the metalation represents the penultimate reaction step in the prokaryotic coproporphyrin-dependent heme biosynthetic pathway, yielding coproheme (ferric coproporphyrin III). By following the metal titration with resonance Raman spectroscopy and x-ray crystallography, we prove that upon metalation the saddling distortion becomes predominant both in the crystal and in solution. This is a consequence of the readjustment of hydrogen bond interactions of the propionates with the protein scaffold during the enzymatic catalysis. Once the propionates have established the interactions typical of the coproheme complex, the distortion slowly decreases, to reach the almost planar final product.


Subject(s)
Coproporphyrins , Iron , Coproporphyrins/metabolism , Iron/metabolism , Ferrochelatase/chemistry , Ferrochelatase/metabolism , Propionates/chemistry , Catalysis
3.
Protein Sci ; 32(1): e4534, 2023 01.
Article in English | MEDLINE | ID: mdl-36479958

ABSTRACT

Coproporphyrin ferrochelatases (CpfCs) are enzymes catalyzing the penultimate step in the coproporphyrin-dependent (CPD) heme biosynthesis pathway, which is mainly utilized by monoderm bacteria. Ferrochelatases insert ferrous iron into a porphyrin macrocycle and have been studied for many decades, nevertheless many mechanistic questions remain unanswered to date. Especially CpfCs, which are found in the CPD pathway, are currently in the spotlight of research. This pathway was identified in 2015 and revealed that the correct substrate for these ferrochelatases is coproporphyrin III (cpIII) instead of protoporphyrin IX, as believed prior the discovery of the CPD pathway. The chemistry of cpIII, which has four propionates, differs significantly from protoporphyrin IX, which features two propionate and two vinyl groups. These findings let us to thoroughly describe the physiological cpIII-ferrochelatase complex in solution and in the crystal phase. Here, we present the first crystallographic structure of the CpfC from the representative monoderm pathogen Listeria monocytogenes bound to its physiological substrate, cpIII, together with the in-solution data obtained by resonance Raman and UV-vis spectroscopy, for wild-type ferrochelatase and variants, analyzing propionate interactions. The results allow us to evaluate the porphyrin distortion and provide an in-depth characterization of the catalytically-relevant binding mode of cpIII prior to iron insertion. Our findings are discussed in the light of the observed structural restraints and necessities for this porphyrin-enzyme complex to catalyze the iron insertion process. Knowledge about this initial situation is essential for understanding the preconditions for iron insertion in CpfCs and builds the basis for future studies.


Subject(s)
Porphyrins , Porphyrins/chemistry , Coproporphyrins/metabolism , Propionates , Catalytic Domain , Ferrochelatase/genetics , Ferrochelatase/chemistry , Ferrochelatase/metabolism , Binding Sites , Iron/metabolism
4.
Biochemistry ; 60(15): 1226-1241, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33784066

ABSTRACT

The catalytic activity of dye-decolorizing peroxidases (DyPs) toward bulky substrates, including anthraquinone dyes, phenolic lignin model compounds, or 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), is in strong contrast to their sterically restrictive active site. In two of the three known subfamilies (A- and C/D-type DyPs), catalytic protein radicals at surface-exposed sites, which are connected to the heme cofactor by electron transfer path(s), have been identified. So far in B-type DyPs, there has been no evidence for protein radical formation after activation by hydrogen peroxide. Interestingly, B-type Klebsiella pneumoniae dye-decolorizing peroxidase (KpDyP) displays a persistent organic radical in the resting state composed of two species that can be distinguished by W-band electron spin echo electron paramagnetic resonance (EPR) spectroscopy. Here, on the basis of a comprehensive mutational and EPR study of computationally predicted tyrosine and tryptophan variants of KpDyP, we demonstrate the formation of tyrosyl radicals (Y247 and Y92) and a radical-stabilizing Y-W dyad between Y247 and W18 in KpDyP, which are unique to enterobacterial B-type DyPs. Y247 is connected to Y92 by a hydrogen bonding network, is solvent accessible in simulations, and is involved in ABTS oxidation. This suggests the existence of long-range electron path(s) in B-type DyPs. The mechanistic and physiological relevance of the reaction mechanism of B-type DyPs is discussed.


Subject(s)
Coloring Agents/metabolism , Electron Spin Resonance Spectroscopy , Models, Molecular , Peroxidases/chemistry , Peroxidases/metabolism , Tyrosine , Color , Electron Transport , Free Radicals/chemistry , Protein Conformation
5.
Molecules ; 25(3)2020 Jan 27.
Article in English | MEDLINE | ID: mdl-32012739

ABSTRACT

The aim of this study was to analyze the binding interactions between a common antihypertensive drug (ramipril, R) and the widely distributed plant flavonoid quercetin (Q), in the presence of human serum albumin (HSA). From the observed fluorescence spectra of the (HSA + R) system we can assume that ramipril is also one of the Site 3 ligands-similar to fusidic acid-the binding of which has been proven by RTG crystallography. Our claim is supported by near-UV CD spectroscopy, microscale themophoresis and molecular modeling. The presence of R slightly inhibited the subsequent binding of Q to HSA and, on the contrary, the pre-incubation of HSA with Q caused a stronger binding of R, most likely due to allosteric interactions. At high concentrations, R is also able to displace Q from its binding site. The dissociation constant KD for the binding of R is more than hundredfold larger than for Q, which means that R is a very weak binder to HSA. The knowledge of qualitative and quantitative parameters of R, as well as the methods used in this study, are important for future research into HSA binding. This study shows the importance of implementing other methods for KD determination. Microscale thermophoresis has proved to be a novel, practical and accurate method for KD determination on HSA, especially in cases when fluorescence spectroscopy is unable to produce usable results.


Subject(s)
Quercetin/metabolism , Ramipril/metabolism , Serum Albumin, Human/metabolism , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Conformation , Quercetin/chemistry , Ramipril/chemistry , Serum Albumin, Human/chemistry
6.
Molecules ; 24(3)2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30704023

ABSTRACT

The aim of this study was to analyze the binding interactions between a common antihypertensive drug (amlodipine besylate-AML) and the widely distributed plant flavonoid quercetin (Q), in the presence of human serum albumin (HSA). Fluorescence analysis was implemented to investigate the effect of ligands on albumin intrinsic fluorescence and to define the binding and quenching properties. Further methods, such as circular dichroism and FT-IR, were used to obtain more details. The data show that both of these compounds bind to Sudlow's Site 1 on HSA and that there exists a competitive interaction between them. Q is able to displace AML from its binding site and the presence of AML makes it easier for Q to bind. AML binds with the lower affinity and if the binding site is already occupied by Q, it binds to the secondary binding site inside the same hydrophobic pocket of Sudlow's Site 1, with exactly the same affinity. Experimental data were complemented with molecular docking studies. The obtained results provide useful information about possible pharmacokinetic interactions upon simultaneous co-administration of the food/dietary supplement and the antihypertensive drug.


Subject(s)
Amlodipine/chemistry , Quantitative Structure-Activity Relationship , Quercetin/chemistry , Serum Albumin, Human/chemistry , Amlodipine/metabolism , Amlodipine/pharmacokinetics , Drug Interactions , Humans , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Quercetin/metabolism , Quercetin/pharmacokinetics , Serum Albumin, Human/metabolism , Spectrum Analysis
7.
Luminescence ; 32(7): 1203-1211, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28513070

ABSTRACT

Protein-binding interactions are displacement reactions which have been implicated as the causative mechanisms in many drug-drug interactions. Thus, the aim of presented study was to analyse human serum albumin-binding displacement interaction between two ligands, hypoglycaemic drug gliclazide and widely distributed plant flavonoid quercetin. Fluorescence analysis was used in order to investigate the effect of substances on intrinsic fluorescence of human serum albumin (HSA) and to define binding and quenching properties of ligand-albumin complexes in binary and ternary systems, respectively. Both ligands showed the ability to bind to HSA, although to a different extent. The displacement effect of one ligand from HSA by the other one has been described on the basis of the quenching curves and binding constants comparison for the binary and ternary systems. According to the fluorescence data analysis, gliclazide presents a substance with a lower binding capacity towards HSA compared with quercetin. Results also showed that the presence of quercetin hindered the interaction between HSA and gliclazide, as the binding constant for gliclazide in the ternary system was remarkably lower compared with the binary system. This finding indicates a possibility for an increase in the non-bound fraction of gliclazide which can lead to its more significant hypoglycaemic effect. Additionally, secondary and tertiary structure conformational alterations of HSA upon binding of both ligands were investigated using synchronous fluorescence, circular dichroism and FT-IR. Experimental data were complemented with molecular docking studies. Obtained results provide beneficial information about possible interference upon simultaneous co-administration of the food/dietary supplement and drug.


Subject(s)
Gliclazide/pharmacology , Quercetin/pharmacology , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Binding, Competitive , Circular Dichroism , Drug Interactions , Gliclazide/metabolism , Molecular Docking Simulation , Protein Conformation , Quercetin/metabolism , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared
8.
Biochem Biophys Rep ; 4: 257-267, 2015 Dec.
Article in English | MEDLINE | ID: mdl-29124212

ABSTRACT

BACKGROUND: The heme enzyme lactoperoxidase is found in body secretions where it significantly contributes to the humoral immune response against pathogens. After activation the peroxidase oxidizes thiocyanate to hypothiocyanite which is known for its microbicidal properties. Yet several pathologies are accompanied by a disturbed hypothiocyanite production which results in a reduced immune defense. METHODS: The results were obtained by measuring enzyme-kinetic parameters using UV-vis spectroscopy and a standardized enzyme-kinetic test system as well as by the determination of second order rate constants using stopped-flow spectroscopy. RESULTS: In this study we systematically tested thirty aromatic substrates for their efficiency to promote the lactoperoxidase-mediated hypothiocyanite production by restoring the native ferric enzyme state. Thereby hydrophobic compounds with a 3,4-dihydroxyphenyl partial structure such as hydroxytyrosol and selected flavonoids emerged as highly efficient promotors of the (pseudo-)halogenating lactoperoxidase activity. CONCLUSIONS: This study discusses important structure-function relationships of efficient aromatic LPO substrates and may contribute to the development of new agents to promote lactoperoxidase activity in secretory fluids of patients. SIGNIFICANCE: This study may contribute to a better understanding of the (patho-)physiological importance of the (pseudo-)halogenating lactoperoxidase activity. The presented results may in future lead to the development of new therapeutic strategies which, by reactivating lactoperoxidase-derived hypothiocyanite production, promote the immunological activity of this enzyme.

9.
J Bacteriol ; 193(10): 2408-17, 2011 May.
Article in English | MEDLINE | ID: mdl-21441524

ABSTRACT

Chlorite dismutase (Cld) is a unique heme enzyme catalyzing the conversion of ClO(2)(-) to Cl(-) and O(2). Cld is usually found in perchlorate- or chlorate-reducing bacteria but was also recently identified in a nitrite-oxidizing bacterium of the genus Nitrospira. Here we characterized a novel Cld-like protein from the chemolithoautotrophic nitrite oxidizer Nitrobacter winogradskyi which is significantly smaller than all previously known chlorite dismutases. Its three-dimensional (3D) crystal structure revealed a dimer of two identical subunits, which sharply contrasts with the penta- or hexameric structures of other chlorite dismutases. Despite a truncated N-terminal domain in each subunit, this novel enzyme turned out to be a highly efficient chlorite dismutase (K(m) = 90 µM; k(cat) = 190 s(-1); k(cat)/K(m) = 2.1 × 10(6) M(-1) s(-1)), demonstrating a greater structural and phylogenetic diversity of these enzymes than was previously known. Based on comparative analyses of Cld sequences and 3D structures, signature amino acid residues that can be employed to assess whether uncharacterized Cld-like proteins may have a high chlorite-dismutating activity were identified. Interestingly, proteins that contain all these signatures and are phylogenetically closely related to the novel-type Cld of N. winogradskyi exist in a large number of other microbes, including other nitrite oxidizers.


Subject(s)
Genetic Variation , Nitrobacter/enzymology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Chlorides/metabolism , Crystallography, X-Ray , Kinetics , Models, Molecular , Nitrobacter/genetics , Nitrobacter/metabolism , Oxidoreductases/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary
10.
J Struct Biol ; 172(3): 331-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20600954

ABSTRACT

Chlorite dismutase (Cld) is a unique heme enzyme which transforms chlorite to chloride and molecular oxygen (reaction: ClO(2)(-)→Cl(-)+O(2)). Since bacteria with Cld play significant roles in the bioremediation of industrially contaminated sites and also in wastewater treatment, it is of high interest to understand the molecular mechanism of chlorite detoxification. Here we investigate a highly active Cld from Candidatus Nitrospira defluvii (NdCld), a key nitrifier in biological wastewater treatment, using a comprehensive structural, biochemical and bioinformatics approach. We determined the crystal structure of Cld from Candidatus Nitrospira defluvii and showed that functional NdCld is a homopentamer possessing a fold found in other Clds and Cld-like enzymes. To investigate the Cld function in more detail, site-directed mutagenesis of a catalytically important residue (Arg173) was performed and two enzyme mutants were structurally and biochemically characterized. Arginine 173 is demonstrated to play a key role in (i) controlling of ligand and substrate access and binding and (ii) in chlorite dismutation reaction. The flexible residue modulates the electrostatic potential and size of the active site entrance and might be involved in keeping transiently formed hypochlorite in place for final molecular oxygen and chloride formation. Furthermore, using a structure-based sequence alignment, we show that the residue corresponding to Arg173 is conserved in all known active forms of Cld and propose it as a marker for Cld activity in yet uncharacterized Cld-like proteins. Finally, our analysis indicates that all Clds and Cld-like enzymes employ a non-covalently bound heme as a cofactor.


Subject(s)
Bacteria/enzymology , Nitrites/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Catalytic Domain , Mutagenesis, Site-Directed , Oxidoreductases/classification , Oxidoreductases/genetics , Protein Structure, Secondary
11.
Arch Biochem Biophys ; 495(1): 21-7, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20018169

ABSTRACT

The heme-containing enzyme myeloperoxidase (MPO) accumulates at inflammatory sites and is able to catalyse one- and two-electron oxidation reactions. Here it is shown that (-)-epicatechin, which is known to have numerous beneficial health effects, in low micromolar concentration enhances the degradation of monochlorodimedon (MCD) or the chlorination of taurine in a concentration-dependent bell-shaped manner whereas at higher concentrations it sufficiently suppresses the release of hypochlorous acid. Presented reaction mechanisms demonstrate the efficiency of micromolar concentrations of the flavan-3-ol in overcoming the accumulation of compound II that does not participate in the chlorination cycle. In case of MCD the mechanism is more complicated since it also acts as peroxidase substrate with very different reactivity towards compound I (3 x 10(5) M(-1) s(-1)) and compound II (8.8M(-1)s(-1)) at pH 7. By affecting the chlorinating activity of myeloperoxidase (-)-epicatechin may participate in regulation of immune responses at inflammatory sites.


Subject(s)
Catechin/pharmacology , Cyclohexanones/metabolism , Peroxidase/metabolism , Taurine/analogs & derivatives , Halogenation/drug effects , Humans , Taurine/metabolism
12.
J Am Chem Soc ; 131(32): 11288-9, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19627079

ABSTRACT

Cytochrome c (cyt c), a mitochondrial intermembrane electron shuttle between complexes III and IV, can, upon binding with an anionic phospholipid, cardiolipin (CL), act as a peroxidase that catalyzes cardiolipin oxidation. H(2)O(2) was considered as a source of oxidative equivalents for this reaction, which is essential for programmed cell death. Here we report that peroxidase cyt c/CL complexes can utilize free fatty acid hydroperoxides (FFA-OOH) at exceptionally high rates that are approximately 3 orders of magnitude higher than for H(2)O(2). Similarly, peroxidase activity of murine liver mitochondria was high with FFA-OOH. Using EPR spin trapping and LC-MS techniques, we have demonstrated that cyt c/CL complexes split FFA-OOH predominantly via a heterolytic mechanism, yielding hydroxy-fatty acids, whereas H(2)O(2) (and tert-butyl hydroperoxide, t-BuOOH) undergo homolytic cleavage. Computer simulations have revealed that Arg(38) and His(33) are important for the heterolytic mechanism at potential FFA-OOH binding sites of cyt c (but not for H(2)O(2) or t-BuOOH). Regulation of FFA-OOH metabolism may be an important function of cyt c that is associated with elimination of toxic FFA-OOH and synthesis of physiologically active hydroxy-fatty acids in mitochondria.


Subject(s)
Antioxidants/metabolism , Cardiolipins/metabolism , Cytochromes c/metabolism , Fatty Acids/metabolism , Hydrogen Peroxide/metabolism , Mitochondria, Liver/enzymology , Animals , Armoracia/enzymology , Models, Molecular , Murinae , Oxidation-Reduction , Protein Binding
13.
Biochem Biophys Res Commun ; 371(4): 810-3, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18466756

ABSTRACT

Apocynin has been reported to require dimerization by myeloperoxidase (MPO) to inhibit leukocyte NADPH oxidase. (-)-Epicatechin, a dietary flavan-3-ol, has been identified as a 'prodrug' of apocynin-like metabolites that inhibit endothelial NADPH oxidase activity and elevate the cellular level of nitric oxide. Since (-)-epicatechin has tentatively been identified as substrate of MPO, we studied the one-electron oxidation of (-)-epicatechin by MPO. By using multi-mixing stopped-flow technique, we demonstrate that (-)-epicatechin is one of the most efficient electron donors for heme peroxidases investigated so far. Second order rate constants for the (-)-epicatechin-mediated conversion of MPO-compound I to compound II and compound II to resting enzyme were estimated to be 1.9 x 10(7) and 4.5 x 10(6) M(-1)s(-1), respectively (pH 7, 25 degrees C). The data indicate that (-)-epicatechin is capable of undergoing fast MPO-mediated one-electron oxidation.


Subject(s)
Catechin/chemistry , Peroxidase/chemistry , Electrons , Humans , Kinetics , Oxidation-Reduction
14.
Biochem Biophys Res Commun ; 356(2): 450-6, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17359937

ABSTRACT

In human myeloperoxidase (MPO) the heme is covalently attached to the protein via two ester linkages and a unique sulfonium ion linkage between the sulfur atom of Met243 and the beta-carbon of the vinyl ring on pyrrole ring A. Here, we have investigated the variant Met243Val produced in Chinese hamster ovary cells in order to elucidate the role of the electron withdrawing sulfonium bond in compound I formation and reduction. Disruption of this MPO-typical bond causes a blue-shifted UV-vis spectrum and an increase in the heme flexibility. This had no impact on compound I formation mediated by hydrogen peroxide (2.2x10(7) M(-1)s(-1) at pH 7.0 and 25 degrees C). Compared with wild-type recombinant MPO the cyanide association rate with ferric Met243Val was significantly enhanced as were also the calculated apparent bimolecular compound I reduction rates by iodide (>10(8) M(-1)s(-1)) and thiocyanate (>10(8) M(-1)s(-1)). By contrast, the overall chlorination and bromination activities were decreased by 98.1% and 87.4%, respectively, compared with the wild-type protein. Compound I reduction by chloride was slower than compound I decay to a compound II-like species (0.4 s(-1)), whereas compound I reduction by bromide was about 10-times slower (1.3x10(4) M(-1)s(-1)) than the wild-type rate. These findings are discussed with respect to the known crystal structure of MPO and its bromide complex as well as the known redox chemistry of its intermediates and substrates.


Subject(s)
Halogens/chemistry , Peroxidase/chemistry , Sulfones/chemistry , Thiocyanates/chemistry , Amino Acid Substitution , Animals , Humans , Methionine/genetics , Oxidation-Reduction , Peroxidase/genetics , Sulfonium Compounds/chemistry , Valine/genetics
15.
J Biol Chem ; 280(51): 42411-22, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16244360

ABSTRACT

Catalase-peroxidases (KatG) are bifunctional heme peroxidases with an overwhelming catalatic activity. The structures show that the buried heme b is connected to the exterior of the enzyme by a main channel built up by KatG-specific loops named large loop LL1 and LL2, the former containing the highly conserved sequence Met-Gly-Leu-Ile-Tyr-Val-Asn-Pro-Glu-Gly. LL1 residues Ile248, Asn251, Pro252, and Glu253 of KatG from Synechocystis are the focus of this study because of their exposure to the solute matrix of the access channel. In particular, the I248F, N251L, P252A, E253Q, and E253D mutants have been analyzed by UV-visible and resonance Raman spectroscopies in combination with steady-state and presteady-state kinetic analyses. Exchange of these residues did not alter the kinetics of cyanide binding or the overall peroxidase activity. Moreover, the kinetics of compound I formation and reduction by one-electron donors was similar in the variants and the wild-type enzyme. However, the turnover numbers of the catalase activity of I248F, N251L, E253Q, and E253D were only 12.3, 32.6, 25, and 42% of the wild-type activity, respectively. These findings demonstrate that the oxidation reaction of hydrogen peroxide (not its reduction) was affected by these mutations. The altered kinetics allowed us to monitor the spectral features of the dominating redox intermediate of E253Q in the catalase cycle. Resonance Raman data and structural analysis demonstrated the existence of a very rigid and ordered structure built up by the interactions of these residues with distal side and also (via LL1) proximal side amino acids, with the heme itself, and with the solute matrix in the channel. The role of Glu253 and the other investigated channel residues in maintaining an ordered matrix of oriented water dipoles, which guides hydrogen peroxide to its site of oxidation, is discussed.


Subject(s)
Catalase/physiology , Peroxidases/physiology , Amino Acid Sequence , Catalase/chemistry , Catalase/genetics , Catalysis , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis , Peroxidases/chemistry , Peroxidases/genetics , Protein Conformation , Sequence Homology, Amino Acid , Spectrum Analysis, Raman
16.
Biochem Biophys Res Commun ; 337(3): 944-54, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16214107

ABSTRACT

Nitric oxide-derived oxidants (e.g., peroxynitrite) are believed to participate in antimicrobial activities as part of normal host defenses but also in oxidative tissue injury in inflammatory disorders. A similar role is ascribed to the heme enzyme myeloperoxidase (MPO), the most abundant protein of polymorphonuclear leukocytes, which are the terminal phagocytosing effector cells of the innate immune system. Concomitant production of peroxynitrite and release of millimolar MPO are characteristic events during phagocytosis. In order to understand the mode of interaction between MPO and peroxynitrite, we have performed a comprehensive stopped-flow investigation of the reaction between all physiological relevant redox intermediates of MPO and peroxynitrite. Both iron(III) MPO and iron(II) MPO are rapidly converted to compound II by peroxynitrite in monophasic reactions with calculated rate constants of (6.8+/-0.1) x 10(6) M(-1)s(-1) and (1.3+/-0.2) x 10(6) M(-1)s(-1), respectively (pH 7.0 and 25 degrees C). Besides these one- and two-electron reduction reactions of peroxynitrite, which produce nitrogen dioxide and nitrite, a one-electron oxidation to the oxoperoxonitrogen radical must occur in the fast monophasic transition of compound I to compound II mediated by peroxynitrite at pH 7.0 [(7.6+/-0.1) x 10(6) M(-1)s(-1)]. In addition, peroxynitrite induced a steady-state transition from compound III to compound II with a rate of (1.0+/-0.3) x 10(4) M(-1)s(-1). Thus, the interconversion among the various oxidation states of MPO that is prompted by peroxynitrite is remarkable. Reaction mechanisms are proposed and the physiological relevance is discussed.


Subject(s)
Flow Injection Analysis/methods , Iron/chemistry , Nitrogen Oxides/chemistry , Peroxidase/chemistry , Peroxynitrous Acid/chemistry , Iron/analysis , Nitrogen Oxides/analysis , Oxidation-Reduction , Peroxidase/analysis , Peroxynitrous Acid/analysis
17.
Biochem Biophys Res Commun ; 332(3): 837-44, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15913556

ABSTRACT

Hypochlorous acid (HOCl) is the most powerful oxidant produced by human neutrophils and contributes to the damage caused by these inflammatory cells. It is produced from H2O2 and chloride by the heme enzyme myeloperoxidase (MPO). Based on findings that betalains provide antioxidant and anti-inflammatory effects, we performed the present kinetic study on the interaction between the betalains, betanin and indicaxanthin, with the redox intermediates, compound I and compound II of MPO, and its major cytotoxic product HOCl. It is shown that both betalains are good peroxidase substrates for MPO and function as one-electron reductants of its redox intermediates, compound I and compound II. Compound I is reduced to compound II with a second-order rate constant of (1.5+/-0.1) x 10(6) M(-1) s(-1) (betanin) and (1.1+/-0.2) x 10(6) M(-1) s(-1) (indicaxanthin), respectively, at pH 7.0 and 25 degrees C. Formation of ferric (native) MPO from compound II occurs with a second-order rate constant of (1.1+/-0.1) x 10(5) M(-1) s(-1) (betanin) and (2.9+/-0.1) x 10(5) M(-1) s(-1) (indicaxanthin), respectively. In addition, both betalains can effectively scavenge hypochlorous acid with determined rates of (1.8+/-0.2) x 10(4) M(-1) s(-1) (betanin) and (7.7+/-0.1) x 10(4) M(-1) s(-1) (indicaxanthin) at pH 7.0 and 25 degrees C. At neutral pH and depending on their concentration, both betalains can exhibit a stimulating and inhibitory effect on the chlorination activity of MPO, whereas at pH 5.0 only inhibitory effects were observed even at micromolar concentrations. These findings are discussed with respect to our knowledge of the enzymatic mechanisms of MPO.


Subject(s)
Hypochlorous Acid/toxicity , Indoles/metabolism , Peroxidase/metabolism , Pyridines/metabolism , Antioxidants/metabolism , Betacyanins , Betaxanthins , Humans , Hypochlorous Acid/metabolism , In Vitro Techniques , Indoles/pharmacology , Inflammation Mediators/metabolism , Kinetics , Oxidants/toxicity , Oxidation-Reduction , Peroxidase/antagonists & inhibitors , Pyridines/pharmacology , Substrate Specificity
18.
Biochemistry ; 44(17): 6482-91, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15850382

ABSTRACT

In human myeloperoxidase the heme is covalently attached to the protein via two ester linkages between the carboxyl groups of Glu242 and Asp94 and modified methyl groups on pyrrole rings A and C of the heme as well as a sulfonium ion linkage between the sulfur atom of Met243 and the beta-carbon of the vinyl group on pyrrole ring A. In the present study, wild-type recombinant myeloperoxidase (recMPO) and the variant Glu242Gln were produced in Chinese hamster ovary cells and investigated in a comparative sequential-mixing stopped-flow study in order to elucidate the role of the Glu242-heme ester linkage in the individual reaction steps of both the halogenation and peroxidase cycle. Disruption of the ester bond increased heme flexibility, blue shifted the UV-vis spectrum, and, compared with recMPO, decelerated cyanide binding (1.25 x 10(4) versus 1.6 x 10(6) M(-)(1) s(-)(1) at pH 7 and 25 degrees C) as well as compound I formation mediated by either hydrogen peroxide (7.8 x 10(5) versus 1.9 x 10(7) M(-)(1) s(-)(1)) or hypochlorous acid (7.5 x 10(5) versus 2.3 x 10(7) M(-)(1) s(-)(1)). The overall chlorination and bromination activity of Glu242Gln was 2.0% and 24% of recMPO. The apparent bimolecular rate constants of compound I reduction by chloride (65 M(-)(1) s(-)(1)), bromide (5.4 x 10(4) M(-)(1) s(-)(1)), iodide (6.4 x 10(5) M(-)(1) s(-)(1)), and thiocyanate (2.2 x10(5) M(-)(1) s(-)(1)) were 500, 25, 21, and 63 times decreased compared with recMPO. By contrast, Glu242Gln compound I reduction by tyrosine was only 5.4 times decreased, whereas tyrosine-mediated compound II reduction was 60 times slower compared with recMPO. The effects of exchange of Glu242 on electron transfer reactions are discussed.


Subject(s)
Glutamic Acid/chemistry , Heme/chemistry , Peroxidase/chemistry , Peroxidase/metabolism , Animals , Aspartic Acid/metabolism , Binding Sites/genetics , Bromides/metabolism , CHO Cells , Chlorides/metabolism , Circular Dichroism , Cricetinae , Cyanides/chemistry , Enzyme Stability/genetics , Eosinophil Peroxidase/metabolism , Ferric Compounds/chemistry , Glutamic Acid/genetics , Glutamine/genetics , Heme/metabolism , Humans , Methionine/metabolism , Oxidation-Reduction , Peroxidase/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
J Inorg Biochem ; 99(5): 1220-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15833345

ABSTRACT

Lactoperoxidase (LPO) is found in mucosal surfaces and exocrine secretions including milk, tears and saliva and has physiological significance in antimicrobial defense. Its predominant physiological role is to convert hydrogen peroxide and thiocyanate in hypothiocyanite. In this study, the standard reduction potentials of all redox couples involved in the halogenation and peroxidase cycle of LPO have been determined by multi-mixing stopped-flow spectroscopy. The standard reduction potentials of the redox couples compound I/native LPO, compound I/compound II of LPO, and compound II/native LPO are (1.09 +/- 0.01) V, (1.14 +/- 0.02) V, and (1.04 +/- 0.02) V, respectively, at pH 7 and 25 degrees C. Thus, for the first time, a full description of these important thermodynamic parameters of lactoperoxidase has been performed, allowing a better understanding in the substantial differences in the oxidation of two- and one-electron donors by LPO and other members of the mammalian heme peroxidase superfamily.


Subject(s)
Lactoperoxidase/chemistry , Animals , Cattle , Lactoperoxidase/metabolism , Milk/enzymology , Oxidation-Reduction
20.
Biochem Pharmacol ; 69(8): 1149-57, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15794935

ABSTRACT

Myeloperoxidase plays a fundamental role in oxidant production by neutrophils. It uses hydrogen peroxide and chloride to catalyze the production of hypochlorous acid (HOCl), which contributes to both bacterial killing and oxidative injury of host tissue. Thus, MPO is an interesting target for anti-inflammatory therapy. Here, based on the extraordinary and MPO-specific redox properties of its intermediates compound I and compound II, we present a rational approach in selection and design of reversible inhibitors of HOCl production mediated by MPO. In detail, indole and tryptamine derivatives were investigated for their ability to reduce compounds I and II and to affect the chlorinating activity of MPO. It is shown that these aromatic one-electron donors bound to the hydrophobic pocket at the distal heme cavity and were oxidized efficiently by compound I (k3), which has a one-electron reduction potential of 1.35 V. By contrast, compound II (E degrees ' of the compound II/ferric couple is 0.97 V) reduction (k4) was extremely slow. As a consequence compound II, which does not participate in the halogenation cycle, accumulated. The extent of chlorinating activity inhibition (IC50) was related to the k3/k4 ratio. The most efficient inhibitors were 5-fluorotryptamine and 5-chlorotryptamine with IC50 of 0.79 microM and 0.73 microM and k3/k4 ratios of 386,000 and 224,000, respectively. The reversible mechanism of inhibition is discussed with respect to the enzymology of MPO and the development of drugs against HOCl-dependent tissue damage.


Subject(s)
Chlorides/metabolism , Enzyme Inhibitors/metabolism , Indoles/pharmacology , Peroxidase/metabolism , Thermodynamics , Tryptamines/pharmacology , Binding Sites , Catalysis , Dose-Response Relationship, Drug , Electrons , Humans , Hydrogen Peroxide/metabolism , Hypochlorous Acid/metabolism , Indoles/chemistry , Indoles/metabolism , Inhibitory Concentration 50 , Kinetics , Molecular Structure , Oxidation-Reduction , Polarography , Spectrophotometry , Structure-Activity Relationship , Substrate Specificity , Tryptamines/chemistry , Tryptamines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...